Elsevier

Bone

Volume 40, Issue 4, April 2007, Pages 981-990
Bone

DU145 human prostate cancer cells express functional receptor activator of NFκB: New insights in the prostate cancer bone metastasis process

https://doi.org/10.1016/j.bone.2006.11.006Get rights and content

Abstract

Prostate cancer metastases to bone are observed in around 80% of prostate cancer patients and represent the most critical complication of advanced prostate cancer, frequently resulting in significant morbidity and mortality. As the underlying mechanisms are not fully characterized, understanding the biological mechanisms that govern prostate cancer metastases to bone at the molecular level should lead to the determination of new potential therapeutic targets. Receptor activator of NFκB ligand (RANKL)/RANK/Osteoprotegerin (OPG) are the key regulators of bone metabolism both in normal and pathological condition, including prostate cancer bone metastases. In the present study, we demonstrated that human prostate cancer cell lines, DU145 and PC3 express biologically functional RANK. Indeed, soluble human RANKL (shRANKL, 100 ng/ml) treatment induced ERK 1/2, p38 and IκB phosphorylations in these cells. shRANKL administration also promoted DU145 and PC3 prostate cancer cell invasion in vitro. Whereas human OPG (hOPG) administration alone (100 ng/ml) had no marked effect, combined association of both agents abolished the RANKL-induced DU145 cell invasion. As RANKL had no direct effect on DU145 cell proliferation, the observed effects were indeed related to RANKL-induced cell migration. DU145 human prostate cancer cells promoted osteoclastogenesis of osteoclast precursors generated from mouse bone marrow. Moreover, DU145 cells produced soluble factor(s) that up-regulate the proliferation of MC3T3-E1 pre-osteoblasts through the activation of the ERK 1/2 and STAT3 signal transduction pathways. This stimulation of pre-osteoblast proliferation resulted in an increased local RANKL expression that can activate both osteoclasts/osteoclast precursors and prostate cancer cells, thus facilitating prostate cancer metastasis development in bone. We confirm that RANKL is a factor that facilitates metastasis to bone by acting as an activator of both osteoclasts and RANK-positive prostate cancer cells in our model. Furthermore, the present study provides the evidence that blocking RANKL–RANK interaction offer new therapeutic approach not only at the level of bone resorbing cells, but also by interfering with RANK-positive prostate cancer cells in the prostate cancer bone metastasis development.

Introduction

Prostate cancer is the most common malignancy diagnosed in males and is currently the major leading cause of cancer death among men [1], [2]. Prostate cancer metastases to bone are observed in around 80% of prostate cancer patients and represent the most critical complication of advanced prostate cancer, frequently resulting in significant morbidity and mortality [1], [2]. Unlike other solid tumors that are associated with osteolytic bone metastases, prostate cancer bone metastases stimulate an overall increase in both bone remodelling and bone volume [3]. In contrast, recent findings strongly suggest the importance of osteoclast function [4], [5], [6], [7], [8], [9]; however the mechanisms underlining these processes are not fully determined. Therefore, understanding the biological mechanisms that govern prostate cancer metastases to bone at the molecular level and elucidation of the interactions among the factors involved should lead to the determination of new potential therapeutic targets.

Receptor activator of NFκB ligand (RANKL)/RANK/Osteoprotegerin (OPG) represent the key regulators of bone metabolism both in normal and pathological condition, including prostate cancer bone metastases [10], [11]. RANKL has been shown to both activate mature osteoclasts and mediate osteoclastogenesis in the presence of macrophage-colony stimulating factor (M-CSF) [12], [13]. RANKL is preferentially expressed on committed pre-osteoblastic cells, whereas its specific receptor RANK is expressed on hematopoietic osteoclast precursors [14], [15]. OPG also produced by osteoblast lineage cells acts as a decoy receptor and inhibits osteoclast formation, function and survival by preventing the binding of RANKL to its receptor RANK [10]. Recent data strongly revealed the significant involvement of the RANKL/RANK/OPG system in metastatic bone cancer diseases, including prostate cancer bone metastases [16]. Indeed, current studies have disclosed that blocking RANKL–RANK interaction prevents the progression of prostate cancer in bone [6], [7], [8], [9]. Furthermore, an increased expression of OPG and RANKL were reported in prostate cancer bone metastases [17], [18]. As OPG is also a decoy receptor for TNF-related apoptosis-inducing ligand (TRAIL), it exerts inhibitory effect on TRAIL-induced cancer cell apoptosis and OPG thus represents a survival factor for prostate cancer cells [19]. RANK expressed on osteoclasts/osteoclast precursors has been largely described as a key receptor that control osteoclast differentiation, activity and survival [12], [14], [15], [20]. However, we and others have demonstrated the expression of functional RANK at the surface of tumor cells that develop in bone, including a mouse osteosarcoma cell line, POS-1 [21], [22]. These recent findings bring new insights in the vicious cycle theory between bone resorbing cells and cancer cells [23]. Indeed, it has been suggested that cancer cells produce soluble factors that activate directly (RANKL) or indirectly via osteoblasts (Parathyroid Hormone-related Protein, Interleukin 8, etc.) osteoclast differentiation and maturation [24]. During the bone resorption, osteoclasts release growth factors stocked in the mineralized bone matrix (Insulin-like Growth Factor-1, Transforming Growth Factor-β, Fibroblast Growth Factor, etc.) that can further activate cancer cell proliferation [25]. This vicious cycle has been proposed to explain the tumor development in bone sites.

Previous studies reported the OPG expression by prostate cancer cells [8] and more recently RANK expression by cancer cells [21], [22], both these findings therefore led us to elucidate the molecular mechanisms underlying prostate cancer bone metastases and the therapeutic relevance of RANKL/RANK/OPG triad in these processes, using DU145 and PC3 human prostate cancer cell lines.

Section snippets

Cell culture

Human prostate cancer cell lines (DU145 and PC3) were obtained from the American Type Culture Collection (ATCC, USA). MC3T3-E1, a mouse calvaria-derived pre-osteoblast cell line was obtained from the RIKEN Cell Bank (Tsukuba, Japan). DU145 and MC3T3-E1 were cultured in Dulbecco's Modified Eagle's Medium (DMEM) (Bio Whittaker, Verviers, Belgium) and PC3 in F12 medium (Invitrogen, Eragny, France), respectively supplemented with 10% fetal bovine serum (FBS, Hyclone, France), at 37 °C in a

Human prostate cancer cell lines DU145 and PC3 express RANK and OPG

The expression of the molecular triad RANKL/RANK/OPG was analyzed in both DU145 and PC3 human prostate cancer cell lines. RT-PCR analyses revealed that both cell lines were positive for RANK and OPG but negative for RANKL at the transcript level (Fig. 1). We further analyzed the expression of major cytokines involved in tumor-bone cells cross-talks through STAT3 signal transduction pathway. Both cell lines were positive for IL-6 and LIF and negative for IL-11 and OSM at the transcript level (

Discussion

The present study was designed to better characterize the involvement of the RANKL–RANK axis in the regulation of bone cells–prostate cancer cells interactions. First, we demonstrated the functional RANK expression at the surface of DU145 and PC3 human prostate cancer cells. The functionality of this receptor was further evidenced as the shRANKL administration promoted human prostate cancer cell invasion in vitro, which was the result of the increased cell migration rather than the increased

Acknowledgments

This work was supported by INSERM, The Région des Pays de la Loire and by a grant from the West Committee of the Ligue Contre le Cancer. Kanji Mori received a personal fellowship from the Ligue Nationale Contre le Cancer.

References (40)

  • C.H. Kim et al.

    Oscillatory fluid flow-induced shear stress decreases osteoclastogenesis through RANKL and OPG signaling

    Bone

    (2006)
  • N. Franchimont et al.

    Interleukin-6: an osteotropic factor influencing bone formation?

    Bone

    (2005)
  • P.C. Smith et al.

    Interleukin-6 and prostate cancer progression

    Cytokine Growth Factor Rev.

    (2001)
  • S.H. Landis et al.

    Cancer statistics, 1999

    CA Cancer J. Clin.

    (1999)
  • G.R. Mundy

    Metastasis to bone: causes, consequences and therapeutic opportunities

    Nat. Rev., Cancer

    (2002)
  • N.W. Clarke et al.

    Morphometric evidence for bone resorption and replacement in prostate cancer

    Br. J. Urol.

    (1991)
  • P. Garnero et al.

    Markers of bone turnover for the management of patients with bone metastases from prostate cancer

    Br. J. Cancer

    (2000)
  • J. Zhang et al.

    Osteoprotegerin inhibits prostate cancer-induced osteoclastogenesis and prevents prostate tumor growth in the bone

    J. Clin. Invest.

    (2001)
  • J. Zhang et al.

    Soluble receptor activator of nuclear factor κB Fc diminishes prostate cancer progression in bone

    Cancer Res.

    (2003)
  • E. Corey et al.

    Osteoprotegerin in prostate cancer bone metastasis

    Cancer Res.

    (2005)
  • Cited by (82)

    • Osteoprotegerin regulates cancer cell migration through SDF-1/CXCR4 axis and promotes tumour development by increasing neovascularization

      2017, Cancer Letters
      Citation Excerpt :

      By its blocking activity of RANKL induced osteoclastogenesis, pre-clinical and clinical studies have investigated the therapeutic potential of recombinant OPG in osteolysis associated with cancer [1]. Preclinical studies demonstrated that the administration of recombinant OPG inhibited osteolysis associated with breast cancer metastasis or multiple myeloma and reduced cancer cell migration in vivo [3]. However, therapeutic use of OPG in bone tumours remains controversial due to its ability to bind and inhibit the TNF related apoptosis inducing ligand (TRAIL) resulting in the inhibition of tumour cells apoptosis [4].

    • Increase of human prostate cancer cell (DU145) apoptosis by telmisartan through PPAR-delta pathway

      2016, European Journal of Pharmacology
      Citation Excerpt :

      The DU145 cell line was originally derived from a human prostate adenocarcinoma metastatic to the brain and prepared by 90 passages in vitro over a period of 2 years (Stone et al., 1978). The cells are not hormone-sensitive and produce soluble factors that activate pre-osteoblast precursors and increase receptor activator of nuclear factor kappa-B ligand (RANKL) expression, thereby facilitating prostate cancer metastasis in bone (Mori et al., 2007). Thus the cell line is a useful model for research on CRPC.

    • Cytokines and bone cancers

      2015, Bone Cancer: Primary Bone Cancers and Bone Metastases: Second Edition
    View all citing articles on Scopus
    View full text